scholarly journals Ex Vivo Isolation and Characterization of Cd4+Cd25+ T Cells with Regulatory Properties from Human Blood

2001 ◽  
Vol 193 (11) ◽  
pp. 1303-1310 ◽  
Author(s):  
Detlef Dieckmann ◽  
Heidi Plottner ◽  
Susanne Berchtold ◽  
Thomas Berger ◽  
Gerold Schuler

It has been known for years that rodents harbor a unique population of CD4+CD25+ “professional” regulatory/suppressor T cells that is crucial for the prevention of spontaneous autoimmune diseases. Here we demonstrate that CD4+CD25+CD45RO+ T cells (mean 6% of CD4+ T cells) are present in the blood of adult healthy volunteers. In contrast to previous reports, these CD4+CD25+ T cells do not constitute conventional memory cells but rather regulatory cells exhibiting properties identical to their rodent counterparts. Cytotoxic T lymphocyte–associated antigen (CTLA)-4 (CD152), for example, which is essential for the in vivo suppressive activity of CD4+CD25+ T cells, was constitutively expressed, and remained strongly upregulated after stimulation. The cells were nonproliferative to stimulation via their T cell receptor for antigen, but the anergic state was partially reversed by interleukin (IL)-2 and IL-15. Upon stimulation with allogeneic (but not syngeneic) mature dendritic cells or platebound anti-CD3 plus anti-CD28 the CD4+CD25+ T cells released IL-10, and in coculture experiments suppressed the activation and proliferation of CD4+ and CD8+ T cells. Suppression proved IL-10 independent, yet contact dependent as in the mouse. The identification of regulatory CD4+CD25+ T cells has important implications for the study of tolerance in man, notably in the context of autoimmunity, transplantation, and cancer.

1998 ◽  
Vol 188 (4) ◽  
pp. 785-790 ◽  
Author(s):  
Jamie D.K. Wilson ◽  
Graham S. Ogg ◽  
Rachel L. Allen ◽  
Philip J.R. Goulder ◽  
Anthony Kelleher ◽  
...  

Acute HIV infection is associated with a vigorous immune response characterized by the proliferation of selected T cell receptor V beta (BV)-expressing CD8+ T cells. These ‘expansions', which are commonly detected in the peripheral blood, can persist during chronic HIV infection and may result in the dominance of particular clones. Such clonal populations are most consistent with antigen-driven expansions of CD8+ T cells. However, due to the difficulties in studying antigen-specific T cells in vivo, it has been hard to prove that oligoclonal BV expansions are actually HIV specific. The use of tetrameric major histocompatibility complex–peptide complexes has recently enabled direct visualization of antigen-specific T cells ex vivo but has not provided information on their clonal composition. We have now made use of these tetrameric complexes in conjunction with anti-BV chain–specific monoclonal antibodies and analysis of cytotoxic T lymphocyte lines/clones to show that chronically clonally expanded CD8+ T cells are HIV specific in vivo.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
David S. Fischer ◽  
Meshal Ansari ◽  
Karolin I. Wagner ◽  
Sebastian Jarosch ◽  
Yiqi Huang ◽  
...  

AbstractThe in vivo phenotypic profile of T cells reactive to severe acute respiratory syndrome (SARS)-CoV-2 antigens remains poorly understood. Conventional methods to detect antigen-reactive T cells require in vitro antigenic re-stimulation or highly individualized peptide-human leukocyte antigen (pHLA) multimers. Here, we use single-cell RNA sequencing to identify and profile SARS-CoV-2-reactive T cells from Coronavirus Disease 2019 (COVID-19) patients. To do so, we induce transcriptional shifts by antigenic stimulation in vitro and take advantage of natural T cell receptor (TCR) sequences of clonally expanded T cells as barcodes for ‘reverse phenotyping’. This allows identification of SARS-CoV-2-reactive TCRs and reveals phenotypic effects introduced by antigen-specific stimulation. We characterize transcriptional signatures of currently and previously activated SARS-CoV-2-reactive T cells, and show correspondence with phenotypes of T cells from the respiratory tract of patients with severe disease in the presence or absence of virus in independent cohorts. Reverse phenotyping is a powerful tool to provide an integrated insight into cellular states of SARS-CoV-2-reactive T cells across tissues and activation states.


F1000Research ◽  
2017 ◽  
Vol 6 ◽  
pp. 1909 ◽  
Author(s):  
D. Branch Moody ◽  
Sara Suliman

The human cluster of differentiation (CD)1 system for antigen display is comprised of four types of antigen-presenting molecules, each with a distinct functional niche: CD1a, CD1b, CD1c, and CD1d. Whereas CD1 proteins were thought solely to influence T-cell responses through display of amphipathic lipids, recent studies emphasize the role of direct contacts between the T-cell receptor and CD1 itself. Moving from molecules to diseases, new research approaches emphasize human CD1-transgenic mouse models and the study of human polyclonal T cells in vivo or ex vivo in disease states. Whereas the high genetic diversity of major histocompatibility complex (MHC)-encoded antigen-presenting molecules provides a major hurdle for designing antigens that activate T cells in all humans, the simple population genetics of the CD1 system offers the prospect of discovering or designing broadly acting immunomodulatory agents.


2021 ◽  
Vol 2 (4) ◽  
pp. 100961
Author(s):  
Clémence Grosjean ◽  
Julie Quessada ◽  
Mathis Nozais ◽  
Marie Loosveld ◽  
Dominique Payet-Bornet ◽  
...  

1998 ◽  
Vol 188 (11) ◽  
pp. 1977-1983 ◽  
Author(s):  
Sally R.M. Bennett ◽  
Francis R. Carbone ◽  
Tracey Toy ◽  
Jacques F.A.P. Miller ◽  
William R. Heath

This report investigates the response of CD8+ T cells to antigens presented by B cells. When C57BL/6 mice were injected with syngeneic B cells coated with the Kb-restricted ovalbumin (OVA) determinant OVA257–264, OVA-specific cytotoxic T lymphocyte (CTL) tolerance was observed. To investigate the mechanism of tolerance induction, in vitro–activated CD8+ T cells from the Kb-restricted, OVA-specific T cell receptor transgenic line OT-I (OT-I cells) were cultured for 15 h with antigen-bearing B cells, and their survival was determined. Antigen recognition led to the killing of the B cells and, surprisingly, to the death of a large proportion of the OT-I CTLs. T cell death involved Fas (CD95), since OT-I cells deficient in CD95 molecules showed preferential survival after recognition of antigen on B cells. To investigate the tolerance mechanism in vivo, naive OT-I T cells were adoptively transferred into normal mice, and these mice were coinjected with antigen-bearing B cells. In this case, OT-I cells proliferated transiently and were then lost from the secondary lymphoid compartment. These data provide the first demonstration that B cells can directly tolerize CD8+ T cells, and suggest that this occurs via CD95-mediated, activation-induced deletion.


1988 ◽  
Vol 11 (3) ◽  
pp. 152-157 ◽  
Author(s):  
Motomu SHIMIZU ◽  
Minako NAKAMURA ◽  
Tateshi KATAOKA ◽  
Takao IWAGUCHI

Blood ◽  
2005 ◽  
Vol 105 (7) ◽  
pp. 2828-2835 ◽  
Author(s):  
Karin Fischer ◽  
Simon Voelkl ◽  
Jana Heymann ◽  
Grzegorz K. Przybylski ◽  
Krishna Mondal ◽  
...  

AbstractDown-regulation of immune responses by regulatory T (Treg) cells is an important mechanism involved in the induction of tolerance to allo-antigens (Ags). Recently, a novel subset of Ag-specific T-cell receptor (TCR)αβ+ CD4-CD8- (double-negative [DN]) Treg cells has been found to be able to prevent the rejection of skin and heart allografts by specifically inhibiting the function of antigraft-specific CD8+ T cells. Here we demonstrate that peripheral DN Treg cells are present in humans, where they constitute about 1% of total CD3+ T cells, and consist of both naïve and Ag-experienced cells. Similar to murine DN Treg cells, human DN Treg cells are able to acquire peptide–HLA-A2 complexes from antigen-presenting cells by cell contact-dependent mechanisms. Furthermore, such acquired peptide-HLA complexes appear to be functionally active, in that CD8+ T cells specific for the HLA-A2–restricted self-peptide, Melan-A, became sensitive to apoptosis by neighboring DN T cells after acquisition of Melan-A–HLA-A2 complexes and revealed a reduced proliferative response. These results demonstrate for the first time that a sizable population of peripheral DN Treg cells, which are able to suppress Ag-specific T cells, exists in humans. DN Treg cells may serve to limit clonal expansion of allo-Ag–specific T cells after transplantation.


Blood ◽  
2018 ◽  
Vol 131 (10) ◽  
pp. 1063-1072 ◽  
Author(s):  
Rupert Handgretinger ◽  
Karin Schilbach

Abstract Allogeneic hematopoetic stem cell transplantation (HCT) offers an option for patients with hematologic malignancies, in whom conventional standard therapies failed or are not effective enough to cure the disease. Successful HCT can restore functional hematopoiesis and immune function, and the new donor-derived immune system can exert a graft-versus-leukemia (GVL) effect. However, allogenic HCT can also be associated with serious risks for transplantation-related morbidities or mortalities such as graft-versus-host disease (GVHD) or life-threatening infectious complications. GVHD is caused by alloreactive T lymphocytes, which express the αβ T-cell receptor, whereas lymphocytes expressing the γδ T-cell receptor are not alloreactive and do not induce GVHD but can exhibit potent antileukemia and anti-infectious activities. Therefore, γδ T cells are becoming increasingly interesting in allogeneic HCT, and clinical strategies to exploit the full function of these lymphocytes have been and are being developed. Such strategies comprise the in vivo activation of γδ T cells or subsets after HCT by certain drugs or antibodies or the ex vivo expansion and manipulation of either patient-derived or donor-derived γδ T cells and their subsets and the adoptive transfer of the ex vivo–activated lymphocytes. On the basis of the absence of dysregulated alloreactivity, such approaches could induce potent GVL effects in the absence of GVHD. The introduction of large-scale clinical methods to enrich, isolate, expand, and manipulate γδ T cells will facilitate future clinical studies that aim to exploit the full function of these beneficial nonalloreactive lymphocytes.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1263-1263
Author(s):  
Jing-Zhou Hou ◽  
Jeanette Baker ◽  
Kelvin Sheehan ◽  
Robert S. Negrin

Abstract Naturally occurring CD4+CD25+ T regulatory cells (Tregs) are a subpopulation of CD4+ T cells vital to homeostasis and maintenance of tolerance. Tregs develop in the thymus and are characterized by expression of the FoxP3 transcription factor. Tregs suppress the proliferation and expansion of conventional CD25-CD4+ and CD8+ T cells through direct cell-cell contact. Defect in Tregs results in multi-organ autoimmune disorders. Adoptive transfer of Tregs cured diabetes in NOD mice and inflammatory bowel disease in murine models. Co-transplantation of Tregs with conventional CD4+ and CD8+ T cells has been shown to control acute graft versus host diseases (GVHD) without abrogating graft versus tumor (GVT) effects in murine bone marrow transplant models.Translating this novel strategy to suppress acute GVHD without loss of GVT holds great promise not only in clinical hematopoietic cell transplantation (HCT), but also in adoptive cellular therapy beyond HCT. Large scale isolation of sufficient human Tregs for therapeutic application remains challenging. Tregs comprise 2–10% of CD4+ T cells. In contrast to rodent Tregs, human CD4+CD25+ cells are more heterogeneous because CD25 is also expressed at lower level by activated non-suppressive conventional T cells and B cells. In addition, immunophenotype of human Tregs is less well studied. Here, we explore the isolation and characterization of human CD4+CD25+ Tregs from peripheral blood mononuclear cells of healthy blood donors and from CD34+ cell depleted fraction of allogenic donors. CD25+ cells were isolated by three different approaches: 1) CD4+ enrichment followed by CD25+ selection, 2) CD19 depletion followed by CD25+ selection, 3) CD25+ selection without prior enrichment or depletion, using magnetic beads. The purities of isolated CD4+CD25+ cells were 95%, 93%, and 93% respectively by FACS. The percentage of CD25 high expressing cells was also similar among the three approaches. More than 90% purity of CD4+CD25+ cells was consistently obtained by CD25+ selection alone using CliniMACS® CD25 microbeads via double column separation utilizing the AutoMACS®. The yield of CD25+ cells varied based upon the ratio of cells/buffer/microbeads. The isolated CD25+ cells were then sorted into CD4+CD25bright (top 25–30%) and CD4+CD25dim (bottom 25–30%) by FACS, fixed and stained for intracellular expression of FoxP3. More than 95% of CD4+CD25bright cells expressed FoxP3. In contrast, only 40% of CD4+CD25dim cells expressed FoxP3. No FoxP3 expression was detected in CD4+CD25− cells. Murine Tregs express a variety of co-stimulatory molecules, however, expression of these molecules is less well characterized on human CD4+CD25+ cells. Using multi-color FACS, we observed that human FoxP3+CD4+CD25bright cells express high levels of CD27 (98%), CD28 (100%), and CD30 (50%) and low levels (<10%) of CTLA-4, GITR, 4-1BB, PD-1 and ICOS on the cell surface. However, CTLA-4, GITR, PD-1 and ICOS were expressed intracellularly in more than 50% of FoxP3+CD4+CD25bright cells. OX40, TRAIL, ICOS ligand and FasL were not detected on the cell surface or in the cytoplasm of FoxP3+CD4+CD25bright cells. FoxP3+CD4+CD25bright cells also express CD62L and CCR5. In summary, highly purified CD4+CD25+ Treg cells can be isolated by magnetic beads and cell sorting. Importantly, the majority of the cells express FoxP3. Utilizing this technique, the clinical application of Tregs appears feasible.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2020-2020
Author(s):  
Eric H. Gschweng ◽  
Melissa N. McCracken ◽  
Thinle Chodon ◽  
Richard C Koya ◽  
Michael L Kaufman ◽  
...  

Abstract Immunotherapy using peripheral blood T-cells engineered with T-cell receptor (TCR) or chimeric antigen receptor genes is a promising approach for the treatment of malignant diseases, and has demonstrated clinical efficacy capable of curing late stage cancer patients. Unfortunately the complete response rate remains low, and the majority of patients respond transiently and then relapse. The massive ex vivo expansion of autologous cells required to generate a therapeutic bolus may exhaust the replicative capacity of the infused cell product. An approach using cells with greater regenerative capacity is an attractive solution to this problem. We hypothesize that gene transfer of an NY-ESO-1 cancer/testes antigen specific TCR into human hematopoietic stem cells (HSCs) is capable of generating a continuous supply of effector T-cells capable of killing cancer in vivo. To evaluate this approach, we utilize a humanized mouse model where peripheral blood stem cells (PBSC) enriched for the stem and progenitor marker CD34+ are transduced with a lentivirus encoding codon optimized NY-ESO-1 TCR and HSV-sr39TK transgenes, then adoptively transferred to preconditioned (100 cGy TBI) NSG-HLA-A2.1 neonatal mice. Development of NY-ESO-1 TCR bearing effector T-cells was detected in the peripheral blood of mice as early as 2 months post-transplant, and persisted for at least 5 months post-transplant. Ex vivo assay of T-cells developed from engineered HSCs showed robust release of interferon-γ when cocultured with NY-ESO-1 antigen and HLA-A2.1 matched tumor cells but not when HLA was mismatched, indicating HLA restricted antigen recognition. Furthermore cytotoxicity assays showed that engineered T-cells were capable of specifically killing tumor cells when antigen and HLA matched. The inclusion of the PET imaging/suicide gene HSV-sr39TK allows both the non-invasive tracking of progeny derived from gene modified HSCs, and their ablation in the event of on-target / off-cancer reactivity or hematopoietic dysplasia due to insertional mutagenesis of gene modified cells. [18F]-FHBG PET imaging of TCR/TK engineered humanized mice allowed the detection of gene modified cells in the marrow compartments of mice, namely the long bones of the legs and arms. Importantly, strong thymus signal was also observed indicating robust thymic population/thymopoiesis of gene modified cells. No specific signal was detected in mock transduced HSC transplanted humanized mice, with identical biodistribution of signal as observed in non-transplanted NSG-A2.1 mice. To examine the suicide gene function of the HSV-sr39TK cassette, we treated engineered humanized mice with vehicle or ganciclovir, the prodrug for suicide gene function of HSV-sr39TK. Ganciclovir treatment resulted in ablation of hematopoietic compartment specific PET signal, while vehicle did not. Mice were subsequently euthanized, and the presence of gene modified cells in organ compartments was examined by digital qPCR for the lentiviral psi element. We detected an order of magnitude decrease in the amount of gene modified cells in ganciclovir treated animals compared with controls. Our studies demonstrate the feasibility of using TCR engineered HSCs for immunotherapy, as functional, HLA-restricted NY-ESO-1 T-cells developed in vivo from transplanted HSCs. In vivo imaging of gene modified cells supports visual resolution of gene modified HSC progeny at a level not previously described, and validation of the suicide gene function of HSV-sr39TK is of particular importance to gene therapy studies. Further work is focused on examining the immunological memory phenotype of TCR modified cells that develop from TCR modified HSCs, the effect of in vivo activation of these cells by dendritic cell pulse, and the ability of NY-ESO-1 humanized mice to mount a persistent anti-tumor response. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document